1
|
Kovecses O, Mercier FE, McKeague M. Nucleic acid therapeutics as differentiation agents for myeloid leukemias. Leukemia 2024; 38:1441-1454. [PMID: 38424137 PMCID: PMC11216999 DOI: 10.1038/s41375-024-02191-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Differentiation therapy has proven to be a success story for patients with acute promyelocytic leukemia. However, the remaining subtypes of acute myeloid leukemia (AML) are treated with cytotoxic chemotherapies that have limited efficacy and a high likelihood of resistance. As differentiation arrest is a hallmark of AML, there is increased interest in developing differentiation-inducing agents to enhance disease-free survival. Here, we provide a comprehensive review of current reports and future avenues of nucleic acid therapeutics for AML, focusing on the use of targeted nucleic acid drugs to promote differentiation. Specifically, we compare and discuss the precision of small interfering RNA, small activating RNA, antisense oligonucleotides, and aptamers to modulate gene expression patterns that drive leukemic cell differentiation. We delve into preclinical and clinical studies that demonstrate the efficacy of nucleic acid-based differentiation therapies to induce leukemic cell maturation and reduce disease burden. By directly influencing the expression of key genes involved in myeloid maturation, nucleic acid therapeutics hold the potential to induce the differentiation of leukemic cells towards a more mature and less aggressive phenotype. Furthermore, we discuss the most critical challenges associated with developing nucleic acid therapeutics for myeloid malignancies. By introducing the progress in the field and identifying future opportunities, we aim to highlight the power of nucleic acid therapeutics in reshaping the landscape of myeloid leukemia treatment.
Collapse
MESH Headings
- Humans
- Cell Differentiation/drug effects
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Nucleic Acids/therapeutic use
- Animals
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/pathology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
- Oligonucleotides, Antisense/therapeutic use
Collapse
Affiliation(s)
- Olivia Kovecses
- Department of Pharmacology and Therapeutics, McGill University, Montreal, H3G 1Y6, QC, Canada
| | - François E Mercier
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, Montreal, H3T 1E2, QC, Canada
| | - Maureen McKeague
- Department of Pharmacology and Therapeutics, McGill University, Montreal, H3G 1Y6, QC, Canada.
- Department of Chemistry, McGill University, Montreal, H3A 0B8, QC, Canada.
| |
Collapse
|
2
|
Thongchot S, Aksonnam K, Thuwajit P, Yenchitsomanus PT, Thuwajit C. Nucleolin‑based targeting strategies in cancer treatment: Focus on cancer immunotherapy (Review). Int J Mol Med 2023; 52:81. [PMID: 37477132 PMCID: PMC10555485 DOI: 10.3892/ijmm.2023.5284] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/15/2023] [Indexed: 07/22/2023] Open
Abstract
The benefits of treating several types of cancers using immunotherapy have recently been established. The overexpression of nucleolin (NCL) in a number of types of cancer provides an attractive antigen target for the development of novel anticancer immunotherapeutic treatments. NCL is a multifunctional protein abundantly distributed in the nucleus, cytoplasm and cell membrane. It influences carcinogenesis, and the proliferation, survival and metastasis of cancer cells, leading to cancer progression. Additionally, the meta‑analysis of total and cytoplasmic NCL overexpression indicates a poor prognosis of patients with breast cancer. The AS1411 aptamers currently appear to have therapeutic action in the phase II clinical trial. The authors' research group has recently explored the anticancer function of NCL through the activation of T cells by dendritic cell‑based immunotherapy. The present review describes and discusses the mechanisms through which the multiple functions of NCL can participate in the progression of cancer. In addition, the studies that define the utility of NCL‑dependent anticancer therapies are summarized, with specific focus being paid to cancer immunotherapeutic approaches.
Collapse
Affiliation(s)
- Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University
| | - Krittaya Aksonnam
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University
| |
Collapse
|
3
|
Mulholland C, Jestřábová I, Sett A, Ondruš M, Sýkorová V, Manzanares CL, Šimončík O, Muller P, Hocek M. The selection of a hydrophobic 7-phenylbutyl-7-deazaadenine-modified DNA aptamer with high binding affinity for the Heat Shock Protein 70. Commun Chem 2023; 6:65. [PMID: 37024672 PMCID: PMC10079658 DOI: 10.1038/s42004-023-00862-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
Nucleic acids aptamers often fail to efficiently target some proteins because of the hydrophilic character of the natural nucleotides. Here we present hydrophobic 7-phenylbutyl-7-deaadenine-modified DNA aptamers against the Heat Shock Protein 70 that were selected via PEX and magnetic bead-based SELEX. After 9 rounds of selection, the pool was sequenced and a number of candidates were identified. Following initial screening, two modified aptamers were chemically synthesised in-house and their binding affinity analysed by two methods, bio-layer interferometry and fluorescent-plate-based binding assay. The binding affinities of the modified aptamers were compared with that of their natural counterparts. The resulting modified aptamers bound with higher affinity (low nanomolar range) to the Hsp70 than their natural sequence (>5 µM) and hence have potential for applications and further development towards Hsp70 diagnostics or even therapeutics.
Collapse
Affiliation(s)
- Catherine Mulholland
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000, Prague 6, Prague, Czech Republic
| | - Ivana Jestřábová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000, Prague 6, Prague, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 8, Prague 2, Prague, 12843, Czech Republic
| | - Arghya Sett
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000, Prague 6, Prague, Czech Republic
| | - Marek Ondruš
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000, Prague 6, Prague, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 8, Prague 2, Prague, 12843, Czech Republic
| | - Veronika Sýkorová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000, Prague 6, Prague, Czech Republic
| | - C Lorena Manzanares
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000, Prague 6, Prague, Czech Republic
- Department of Chemistry and Center for NanoScience, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13 Haus E, 81377, München, Germany
| | - Oliver Šimončík
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute (MMCI), Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Petr Muller
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute (MMCI), Zluty Kopec 7, 656 53, Brno, Czech Republic
| | - Michal Hocek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000, Prague 6, Prague, Czech Republic.
- Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 8, Prague 2, Prague, 12843, Czech Republic.
| |
Collapse
|
4
|
Allen NC, Chauhan R, Bates PJ, O’Toole MG. Optimization of Tumor Targeting Gold Nanoparticles for Glioblastoma Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3869. [PMID: 36364644 PMCID: PMC9653665 DOI: 10.3390/nano12213869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
Glioblastoma brain tumors represent an aggressive form of gliomas that is hallmarked by being extremely invasive and aggressive due to intra and inter-tumoral heterogeneity. This complex tumor microenvironment makes even the newer advancements in glioblastoma treatment less effective long term. In developing newer treatment technologies against glioblastoma, one should tailor the treatment to the tumor microenvironment, thus allowing for a more robust and sustained anti-glioblastoma effect. Here, we present a novel gold nanoparticle therapy explicitly designed for bioactivity against glioblastoma representing U87MG cell lines. We employ standard conjugation techniques to create oligonucleotide-coated gold nanoparticles exhibiting strong anti-glioblastoma behavior and optimize their design to maximize bioactivity against glioblastoma. Resulting nanotherapies are therapy specific and show upwards of 75% inhibition in metabolic and proliferative activity with stark effects on cellular morphology. Ultimately, these gold nanotherapies are a good base for designing more multi-targeted approaches to fighting against glioblastoma.
Collapse
Affiliation(s)
- Nicholas C. Allen
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Rajat Chauhan
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Paula J. Bates
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Martin G. O’Toole
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
5
|
Functional Downregulation of PD-L1 and PD-L2 by CpG and non-CpG Oligonucleotides in Melanoma Cells. Cancers (Basel) 2022; 14:cancers14194698. [PMID: 36230620 PMCID: PMC9562717 DOI: 10.3390/cancers14194698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Although metastatic melanoma is still not a curable disease, targeting of immunologically relevant checkpoints represents a turning point in the treatment. Particularly, targeting the interaction between PD-L1 and its referring receptor PD-1 with antibodies has been shown to activate T-cell function abrogating the evasion of tumor cells from immune recognition. Here, we present another approach that interferes with this system by showing that treatment of melanoma cells with oligonucleotides reduces the expression of PD-L1 (and PD-L2) on tumor cells. Specifically, non-CpG-6-PTO, an ODN that forms superstructures known as G-quartets, has been found to inhibit the interferon-γ-induced signaling cascade which fosters PD-L1 expression. These findings suggest a new therapeutic strategy to interfere with one of the most important immune checkpoints. Abstract The clinical application of immune checkpoint inhibitors represents a breakthrough progress in the treatment of metastasized melanoma and other tumor entities. In the present study, it was hypothesized that oligonucleotides (ODNs), known as modulators of the immune response, have an impact on the endogenous expression of checkpoint molecules, namely PD-L1 and PD-L2 (PD-L1/2). IFNγ-stimulated melanoma cells (A375, SK-Mel-28) were treated with different synthetically manufactured oligonucleotides which differed in sequence, length and backbone composition. It was found that a variety of different ODN sequences significantly suppressed PD-L1/2 expression. This effect was dependent on length and phosphorothioate (PTO) backbone. In particular, a sequence containing solely guanines (nCpG-6-PTO) was highly effective in downregulating PD-L1/2 at the protein, mRNA and promoter levels. Mechanistically, we gave evidence that ODNs with G-quartet-forming motifs suppress the interferon signaling axis (JAK/STAT/IRF1). Our findings identify a subset of ODNs as interesting pharmacological compounds that could expand the arsenal of targeted therapies to combat the immunological escape of tumor cells.
Collapse
|
6
|
Applying Synthetic Biology with Rational Design to Nature’s Greatest Challenges: Bioengineering Immunotherapeutics for the Treatment of Glioblastoma. IMMUNO 2021. [DOI: 10.3390/immuno2010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Improvements in bioengineering methodology and tools have allowed for significant progress in the development of therapeutics and diagnostics in medicine, as well as progress in many other diverse industries, such as materials manufacturing, food and agriculture, and consumer goods. Glioblastomas present significant challenges to adequate treatment, in part due to their immune-evasive and manipulative nature. Rational-design bioengineering using novel scaffolds, biomaterials, and inspiration across disciplines can push the boundaries in treatment development to create effective therapeutics for glioblastomas. In this review, we will discuss bioengineering strategies currently applied across diseases and disciplines to inspire creative development for GBM immunotherapies.
Collapse
|
7
|
Shin C, Kim SS, Jo YH. Extending traditional antibody therapies: Novel discoveries in immunotherapy and clinical applications. Mol Ther Oncolytics 2021; 22:166-179. [PMID: 34514097 PMCID: PMC8416972 DOI: 10.1016/j.omto.2021.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Immunotherapy has been well regarded as one of the safer and antigen-specific anti-cancer treatments compared to first-generation chemotherapy. Since Coley's discovery, researchers focused on engineering novel antibody-based therapies. Including artificial and modified antibodies, such as antibody fragments, antibody-drug conjugates, and synthetic mimetics, the variety of immunotherapy has been rapidly expanding in the last few decades. Genetic and chemical modifications to monoclonal antibody have been brought into academia, in vivo trials, and clinical applications. Here, we have looked around antibodies overall. First, we elucidate the antibody structure and its cytotoxicity mechanisms. Second, types of therapeutic antibodies are presented. Additionally, there is a summarized list of US Food and Drug Administration (FDA)-approved therapeutic antibodies and recent clinical trials. This review provides a comprehensive overview of both the general function of therapeutic antibodies and a few main variations in development, including recent advent with the proposed mechanism of actions, and we introduce types of therapeutic antibodies, clinical trials, and approved commercial immunotherapeutic drugs.
Collapse
Affiliation(s)
- Charles Shin
- Chadwick International, Incheon 22002, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong Hwa Jo
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
8
|
Lopes-Nunes J, Agonia AS, Rosado T, Gallardo E, Palmeira-de-Oliveira R, Palmeira-de-Oliveira A, Martinez-de-Oliveira J, Fonseca-Moutinho J, Campello MPC, Paiva A, Paulo A, Vulgamott A, Ellignton AD, Oliveira PA, Cruz C. Aptamer-Functionalized Gold Nanoparticles for Drug Delivery to Gynecological Carcinoma Cells. Cancers (Basel) 2021; 13:4038. [PMID: 34439193 PMCID: PMC8391588 DOI: 10.3390/cancers13164038] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022] Open
Abstract
Cervical cancer is one of the most common cancers and is one of the major cause of deaths in women, especially in underdeveloped countries. The patients are usually treated with surgery, radiotherapy, and chemotherapy. However, these treatments can cause several side effects and may lead to infertility. Another concerning gynecologic cancer is endometrial cancer, in which a high number of patients present a poor prognosis with low survival rates. AS1411, a DNA aptamer, increases anticancer therapeutic selectivity, and through its conjugation with gold nanoparticles (AS1411-AuNPs) it is possible to improve the anticancer effects. Therefore, AS1411-AuNPs are potential drug carriers for selectively delivering therapeutic drugs to cervical cancer. In this work, we used AS1411-AuNPs as a carrier for an acridine orange derivative (C8) or Imiquimod (IQ). The AS1411 aptamer was covalently bound to AuNPs, and each drug was associated via supramolecular assembly. The final nanoparticles presented suitable properties for pharmaceutical applications, such as small size, negative charge, and favorable drug release properties. Cellular uptake was characterized by confocal microscopy and flow cytometry, and effects on cellular viability were determined by MTT assay. The nanoparticles were then incorporated into a gel formulation of polyethylene glycol, suitable for topical application in the female genital tract. This gel showed promising tissue retention properties in Franz cells studies in the porcine vaginal epithelia. These findings suggest that the tested nanoparticles are promising drug carriers for cervical cancer therapy.
Collapse
Affiliation(s)
- Jessica Lopes-Nunes
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
| | - Ana S. Agonia
- Labfit-HPRD Health Products Research and Development, Lda, Edifício UBIMEDICAL Estrada Municipal 506, 6200-284 Covilhã, Portugal;
| | - Tiago Rosado
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
- C4-Cloud Computing Competence Centre, UBIMedical, Universidade da Beira Interior, EM506, 6200-284 Covilhã, Portugal
| | - Eugénia Gallardo
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
| | - Rita Palmeira-de-Oliveira
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
- Labfit-HPRD Health Products Research and Development, Lda, Edifício UBIMEDICAL Estrada Municipal 506, 6200-284 Covilhã, Portugal;
- Center for Neuroscience and Cell Biology, University of Coimbra, Universidade de Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Ana Palmeira-de-Oliveira
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
- Labfit-HPRD Health Products Research and Development, Lda, Edifício UBIMEDICAL Estrada Municipal 506, 6200-284 Covilhã, Portugal;
| | - José Martinez-de-Oliveira
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
| | - José Fonseca-Moutinho
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
- Quinta do Alvito, Centro Hospitalar Universitário Cova da Beira, 6200-251 Covilhã, Portugal
| | - Maria Paula Cabral Campello
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal; (M.P.C.C.); (A.P.)
- DECN-Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Artur Paiva
- Unidade de Gestão Operacional em Citometria, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-075 Coimbra, Portugal;
- CIMAGO/iCBR/CIBB, Faculdade de Medicina da Universidade de Coimbra, 3000-370 Coimbra, Portugal
- Ciências Biomédicas Laboratoriais, Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, 3046-854 Coimbra, Portugal
| | - António Paulo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal; (M.P.C.C.); (A.P.)
- DECN-Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - Alexa Vulgamott
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA; (A.V.); (A.D.E.)
| | - Andrew D. Ellignton
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA; (A.V.); (A.D.E.)
| | - Paula A. Oliveira
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás os Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Carla Cruz
- CICS-UBI Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.L.-N.); (T.R.); (E.G.); (R.P.-d.-O.); (A.P.-d.-O.); (J.M.-d.-O.); (J.F.-M.)
| |
Collapse
|
9
|
Lopes-Nunes J, Oliveira PA, Cruz C. G-Quadruplex-Based Drug Delivery Systems for Cancer Therapy. Pharmaceuticals (Basel) 2021; 14:671. [PMID: 34358097 PMCID: PMC8308530 DOI: 10.3390/ph14070671] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
G-quadruplexes (G4s) are a class of nucleic acids (DNA and RNA) with single-stranded G-rich sequences. Owing to the selectivity of some G4s, they are emerging as targeting agents to overtake side effects of several potential anticancer drugs, and delivery systems of small molecules to malignant cells, through their high affinity or complementarity to specific targets. Moreover, different systems are being used to improve their potential, such as gold nano-particles or liposomes. Thus, the present review provides relevant data about the different studies with G4s as drug delivery systems and the challenges that must be overcome in the future research.
Collapse
Affiliation(s)
- Jéssica Lopes-Nunes
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal;
| | - Paula A. Oliveira
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Carla Cruz
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal;
| |
Collapse
|
10
|
Carvalho J, Lopes-Nunes J, Vialet B, Rosado T, Gallardo E, Vale J, Eloy C, Ferreira S, Palmeira-de-Oliveira R, Campello MPC, Paulo A, Barthélémy P, Mergny JL, Salgado GF, Queiroz JA, Ellington AD, Cruz C. Nanoaggregate-forming lipid-conjugated AS1411 aptamer as a promising tumor-targeted delivery system of anticancer agents in vitro. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 36:102429. [PMID: 34174419 DOI: 10.1016/j.nano.2021.102429] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/09/2021] [Accepted: 06/06/2021] [Indexed: 11/28/2022]
Abstract
Nanoparticles offer targeted delivery of drugs with minimal toxicity to surrounding healthy tissue and have great potential in the management of human papillomavirus (HPV)-related diseases. We synthesized lipid-modified AS1411 aptamers capable of forming nanoaggregates in solution containing Mg2+. The nanoaggregates presented suitable properties for pharmaceutical applications such as small size (100 nm), negative charge, and drug release. The nanoaggregates were loaded with acridine orange derivative C8 for its specific delivery into cervical cancer cell lines and HPV-positive tissue biopsies. This improved inhibition of HeLa proliferation and cell uptake without significantly affecting healthy cells. Finally, the nanoaggregates were incorporated in a gel formulation with promising tissue retention properties aiming at developing a local delivery strategy of the nanoaggregates in the female genital tract. Collectively, these findings suggest that the nanoformulation protocol has great potential for the delivery of both anticancer and antiviral agents, becoming a novel modality for cervical cancer management.
Collapse
Affiliation(s)
- Josué Carvalho
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Jéssica Lopes-Nunes
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Brune Vialet
- Univ. Bordeaux, CNRS, INSERM, ARNA, UMR 5320, U1212, Bordeaux, France
| | - Tiago Rosado
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal; C4 - Cloud Computing Competence Centre, UBIMedical, Universidade da Beira Interior, EM506, Covilhã, Portugal
| | - Eugenia Gallardo
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - João Vale
- Laboratório de Anatomia Patológica, Ipatimup Diagnósticos, Porto, Portugal
| | - Catarina Eloy
- Laboratório de Anatomia Patológica, Ipatimup Diagnósticos, Porto, Portugal; Faculdade de Medicina, Universidade do Porto, Alameda Prof Hernâni Monteiro, Porto, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Sofia Ferreira
- C4 - Cloud Computing Competence Centre, UBIMedical, Universidade da Beira Interior, EM506, Covilhã, Portugal; Labfit, HPRD - Health Products Research and Development, Lda, Edifício UBIMEDICAL, Covilhã, Portugal
| | - Rita Palmeira-de-Oliveira
- Labfit, HPRD - Health Products Research and Development, Lda, Edifício UBIMEDICAL, Covilhã, Portugal
| | - Maria Paula Cabral Campello
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Portugal; DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | - António Paulo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Portugal; DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | | | - Jean-Louis Mergny
- Institute of Biophysics of the CAS, Brno, Czech Republic; Laboratoire d'Optique et Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, Palaiseau, France
| | - Gilmar F Salgado
- Univ. Bordeaux, CNRS, INSERM, ARNA, UMR 5320, U1212, Bordeaux, France
| | - João A Queiroz
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Andrew D Ellington
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Carla Cruz
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal.
| |
Collapse
|
11
|
Li Z, Fu X, Huang J, Zeng P, Huang Y, Chen X, Liang C. Advances in Screening and Development of Therapeutic Aptamers Against Cancer Cells. Front Cell Dev Biol 2021; 9:662791. [PMID: 34095130 PMCID: PMC8170048 DOI: 10.3389/fcell.2021.662791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/21/2021] [Indexed: 01/10/2023] Open
Abstract
Cancer has become the leading cause of death in recent years. As great advances in medical treatment, emerging therapies of various cancers have been developed. Current treatments include surgery, radiotherapy, chemotherapy, immunotherapy, and targeted therapy. Aptamers are synthetic ssDNA or RNA. They can bind tightly to target molecules due to their unique tertiary structure. It is easy for aptamers to be screened, synthesized, programmed, and chemically modified. Aptamers are emerging targeted drugs that hold great potentials, called therapeutic aptamers. There are few types of therapeutic aptamers that have already been approved by the US Food and Drug Administration (FDA) for disease treatment. Now more and more therapeutic aptamers are in the stage of preclinical research or clinical trials. This review summarized the screening and development of therapeutic aptamers against different types of cancer cells.
Collapse
Affiliation(s)
- Zheng Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xuekun Fu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jie Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Peiyuan Zeng
- Department of Biochemistry, University of Victoria, Victoria, BC, Canada
| | - Yuhong Huang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xinxin Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
12
|
Jin H. Perspectives of Aptamers for Medical Applications. APTAMERS FOR MEDICAL APPLICATIONS 2021:405-462. [DOI: 10.1007/978-981-33-4838-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Lopes-Nunes J, Lifante J, Shen Y, Ximendes EC, Jaque D, Iglesias-de la Cruz MC, Cruz C. Biological studies of an ICG-tagged aptamer as drug delivery system for malignant melanoma. Eur J Pharm Biopharm 2020; 154:228-235. [PMID: 32707287 DOI: 10.1016/j.ejpb.2020.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/20/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Malignant melanoma accounts for about 1% of all skin malignant tumors and represents the most aggressive and lethal form of skin cancer. Clinically, there exist different therapeutic options for melanoma treatment, such as surgery, chemotherapy, radiotherapy, photodynamic therapy and immunotherapy. However, serious adverse effects usually arise, and survival rates are still low because a high number of patients present relapses within 6-9 months after therapy. AS1411 is a G-quadruplex (G4) aptamer capable of tumor-specific recognition, since it binds to nucleolin, a multi-functional protein expressed in many different types of cancer cells. In this work, we present a novel drug delivery system composed of AS1411 and indocyanine green (ICG) to track its accumulation in tumoral cells in a melanoma mouse model. Using a simple supramolecular strategy, we conjugated the complex AS1411-ICG with C8 ligand, an acridine orange derivative with potential anticancer ligand. Then, we performed in vitro cytotoxicity experiments using the B16 mouse melanoma cell line, and in vivo experiments using a B16 mouse melanoma model to study biodistribution and histological changes. The circular dichroism (CD) data suggest that C8 does not affect the parallel G4 topology of AS1411-ICG, whereas it increases its thermal stability. Incubation of B16 melanoma cells with the AS1411-ICG complex associated with C8 increases the cytotoxicity compared with AS1411-ICG alone. From the in vivo studies, we conclude that both AS1411-ICG and AS1411-ICG-C8 presented the potential to accumulate preferentially in tumor tissues. Moreover, these compounds seem to be efficiently removed from the mice's bodies through kidney clearance. In summary, these results suggest that these complexes derived from AS1411 aptamer could act as a delivery system of ligands with antitumoral activity for in vivo melanoma therapy.
Collapse
Affiliation(s)
- Jéssica Lopes-Nunes
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - José Lifante
- Fluorescence Imaging Group, Departamento de Fisiología - Facultad de Medicina, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid 28029, Spain; Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain
| | - Yingli Shen
- Fluorescence Imaging Group, Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain
| | - Erving C Ximendes
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain; Fluorescence Imaging Group, Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain
| | - Daniel Jaque
- Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain; Fluorescence Imaging Group, Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, C/Francisco Tomás y Valiente 7, Madrid 28049, Spain
| | - M Carmen Iglesias-de la Cruz
- Fluorescence Imaging Group, Departamento de Fisiología - Facultad de Medicina, Avda. Arzobispo Morcillo 2, Universidad Autónoma de Madrid, Madrid 28029, Spain; Nanobiology Group, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Ctra. Colmenar km. 9.100, Madrid 28034, Spain
| | - Carla Cruz
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
14
|
Yang Y, Zhao W, Tan W, Lai Z, Fang D, Jiang L, Zuo C, Yang N, Lai Y. An Efficient Cell-Targeting Drug Delivery System Based on Aptamer-Modified Mesoporous Silica Nanoparticles. NANOSCALE RESEARCH LETTERS 2019; 14:390. [PMID: 31872318 PMCID: PMC6928176 DOI: 10.1186/s11671-019-3208-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 11/12/2019] [Indexed: 05/05/2023]
Abstract
How to deliver chemotherapeutic drugs efficiently and selectively to tumor cells to improve therapeutic efficacy remains a difficult problem. We herein construct an efficient cell-targeting drug delivery system (Sgc8-MSN/Dox) based on aptamer-modified mesoporous silica nanoparticles that relies on the tumor-targeting ability of the aptamer Sgc8 to deliver doxorubicin (Dox) to leukemia cells in a targeted way, thereby improving therapeutic efficacy and reducing toxicity. In this work, Sgc8-MSN/Dox showed sustained Dox release, and they targeted and efficiently killed CCRF-CEM human acute T lymphocyte leukemia cells, suggesting potential as a cancer therapy.
Collapse
Affiliation(s)
- Yang Yang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Weihua Zhao
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenwen Tan
- Department of Pharmacy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Zongqiang Lai
- Department of Pharmacy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China
| | - Dong Fang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Lei Jiang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chuantian Zuo
- Department of Surgery Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Nuo Yang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Yongrong Lai
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
15
|
Abstract
This chapter provides a brief introduction to followed by discussion of recent preclinical studies on potential aptamer drugs grouped into two broad categories, namely, “aptamer structures” and “non-ocular diseases.” Examples of aptamer-based targeting of drugs are then described. Next is an overview of the status of nearly 30 clinical trials of aptamer drugs currently listed in ClinicalTrials.gov, which is a registry and results database of publicly and privately supported clinical studies of human participants conducted around the world, and is a service of the US National Institutes of Health. This overview includes brief descriptions of each study sponsor, aptamer drug, disease(s), and type of study, as well as separate tables for completed studies, withdrawn or terminated studies, and active studies. The final section discusses Conclusions and Prospects.
Collapse
Affiliation(s)
- G. Zon
- TriLink BioTechnologies 9955 Mesa Rim Road San Diego 92121 USA
| |
Collapse
|
16
|
Gao G, Liu C, Jain S, Li D, Wang H, Zhao Y, Liu J. Potential use of aptamers for diagnosis and treatment of pancreatic cancer. J Drug Target 2019; 27:853-865. [PMID: 30596288 DOI: 10.1080/1061186x.2018.1564924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pancreatic cancer (PC) is highly malignant with a low 5-year survival rate. PC currently does not have good early diagnostic markers and responses poorly to chemotherapeutic drugs. The search for better biomarkers and developing more effective chemotherapy are important ways to improve the healthcare of PC patients. Aptamers are single-stranded nucleic acids with high binding affinity and specificity to target molecules. Many aptamers against different forms of cancer including PC have been selected for both diagnostic and therapeutic use. Aptamers can work as ligands to distinguish tumour cells from normal cells. Using cells as selection targets, the obtained aptamers have been used to discover new cancer biomarkers after identification of the binding target. Aptamers have been shown to have antagonists effect on cancer cell proliferation, apoptosis, and metastasis. In addition, aptamers have been used as carriers to deliver therapeutic agents to selectively kill PC cells. This review summarises the potential use of aptamers in the diagnosis and treatment of PC.
Collapse
Affiliation(s)
- Ge Gao
- a Faculty of Laboratory Medicine , Xiangya Medical College, Central South University , Changsha , China.,b Department of Clinical Laboratory , Third Xiangya Hospital, Central South University , Changsha , China
| | - Can Liu
- a Faculty of Laboratory Medicine , Xiangya Medical College, Central South University , Changsha , China.,b Department of Clinical Laboratory , Third Xiangya Hospital, Central South University , Changsha , China
| | - Sona Jain
- c Department of Chemistry , Waterloo Institute for Nanotechnology, University of Waterloo , Waterloo , Canada
| | - Dai Li
- c Department of Chemistry , Waterloo Institute for Nanotechnology, University of Waterloo , Waterloo , Canada.,d Department of Pharmacology , Xiangya Hospital, Central South University , Changsha , China
| | - Hai Wang
- a Faculty of Laboratory Medicine , Xiangya Medical College, Central South University , Changsha , China.,b Department of Clinical Laboratory , Third Xiangya Hospital, Central South University , Changsha , China
| | - Yongxin Zhao
- a Faculty of Laboratory Medicine , Xiangya Medical College, Central South University , Changsha , China.,b Department of Clinical Laboratory , Third Xiangya Hospital, Central South University , Changsha , China
| | - Juewen Liu
- c Department of Chemistry , Waterloo Institute for Nanotechnology, University of Waterloo , Waterloo , Canada
| |
Collapse
|
17
|
Tuna BG, Atalay PB, Kuku G, Acar EE, Kara HK, Yilmaz MD, Ozalp VC. Enhanced antitumor activity of carbendazim on HeLa cervical cancer cells by aptamer mediated controlled release. RSC Adv 2019; 9:36005-36010. [PMID: 35540590 PMCID: PMC9074919 DOI: 10.1039/c9ra07974b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/28/2019] [Indexed: 01/13/2023] Open
Abstract
Carbendazim, is a broad-spectrum fungicide and also a promising experimental antitumor drug as reproduction and developmental toxicant, which is currently under phase II preclinical trials. In this study, an approach based on controlled and targeted release with aptamers and mesoporous silica nanoparticles was investigated to improve the antitumor activity of carbendazim. To this end, we synthesized aptamer conjugated silica nanoparticles for testing cytotoxicity properties in vitro with human cervical adenocarcinoma (HeLa) cultured cells. Nucleolin (AS1411) binding aptamers were used to entrap carbendazim molecules inside nanopores of MCM-41 type silica nanoparticles to obtain a stimuli-dependent release system. The effect of carbendazim loaded aptamer silica complex was tested and compared to free carbendazim treatment on HeLa cells, demonstrating 3.3 fold increase of toxicity on targeted cells with our delivery system. In addition, cytotoxicity of the complex was determined to be mostly due to increased apoptosis and to a less extend necrosis related pathways. Carbendazim doped and aptamer-gate functionalized mesoporous silica nanoparticles targeted nucleolin on HeLa cell surface for specific delivery. This delivery system improved antitumor activity of carbendazim by about 3 folds increase of EC50 values.![]()
Collapse
Affiliation(s)
- Bilge G. Tuna
- Department of Biophysics
- Yeditepe University School of Medicine
- Yeditepe University
- Istanbul
- Turkey
| | - Pinar B. Atalay
- Department of Medical Biology and Genetics
- Maltepe University Faculty of Medicine
- Maltepe University
- Istanbul
- Turkey
| | - Gamze Kuku
- Department of Genetics and Engineering
- Faculty of Engineering and Architecture
- Yeditepe University
- Istanbul
- Turkey
| | - E. Esma Acar
- Research and Development Center for Diagnostic Kits (KIT-ARGEM)
- Department of Bioengineering
- Konya Food and Agriculture University
- Konya
- Turkey
| | - H. Kubra Kara
- Research and Development Center for Diagnostic Kits (KIT-ARGEM)
- Department of Bioengineering
- Konya Food and Agriculture University
- Konya
- Turkey
| | - M. Deniz Yilmaz
- Research and Development Center for Diagnostic Kits (KIT-ARGEM)
- Department of Bioengineering
- Konya Food and Agriculture University
- Konya
- Turkey
| | - V. Cengiz Ozalp
- Research and Development Center for Diagnostic Kits (KIT-ARGEM)
- Department of Bioengineering
- Konya Food and Agriculture University
- Konya
- Turkey
| |
Collapse
|
18
|
Bayat P, Nosrati R, Alibolandi M, Rafatpanah H, Abnous K, Khedri M, Ramezani M. SELEX methods on the road to protein targeting with nucleic acid aptamers. Biochimie 2018; 154:132-155. [PMID: 30193856 DOI: 10.1016/j.biochi.2018.09.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/02/2018] [Indexed: 12/14/2022]
Abstract
Systematic evolution of ligand by exponential enrichment (SELEX) is an efficient method used to isolate high-affinity single stranded oligonucleotides from a large random sequence pool. These SELEX-derived oligonucleotides named aptamer, can be selected against a broad spectrum of target molecules including proteins, cells, microorganisms and chemical compounds. Like antibodies, aptamers have a great potential in interacting with and binding to their targets through structural recognition and are therefore called "chemical antibodies". However, aptamers offer advantages over antibodies including smaller size, better tissue penetration, higher thermal stability, lower immunogenicity, easier production, lower cost of synthesis and facilitated conjugation or modification with different functional moieties. Thus, aptamers represent an attractive substitution for protein antibodies in the fields of biomarker discovery, diagnosis, imaging and targeted therapy. Enormous interest in aptamer technology triggered the development of SELEX that has underwent numerous modifications since its introduction in 1990. This review will discuss the recent advances in SELEX methods and their advantages and limitations. Aptamer applications are also briefly outlined in this review.
Collapse
Affiliation(s)
- Payam Bayat
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rahim Nosrati
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Inflammation and Inflammatory Diseases Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Khedri
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Morita Y, Leslie M, Kameyama H, Volk DE, Tanaka T. Aptamer Therapeutics in Cancer: Current and Future. Cancers (Basel) 2018; 10:cancers10030080. [PMID: 29562664 PMCID: PMC5876655 DOI: 10.3390/cancers10030080] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Aptamer-related technologies represent a revolutionary advancement in the capacity to rapidly develop new classes of targeting ligands. Structurally distinct RNA and DNA oligonucleotides, aptamers mimic small, protein-binding molecules and exhibit high binding affinity and selectivity. Although their molecular weight is relatively small—approximately one-tenth that of monoclonal antibodies—their complex tertiary folded structures create sufficient recognition surface area for tight interaction with target molecules. Additionally, unlike antibodies, aptamers can be readily chemically synthesized and modified. In addition, aptamers’ long storage period and low immunogenicity are favorable properties for clinical utility. Due to their flexibility of chemical modification, aptamers are conjugated to other chemical entities including chemotherapeutic agents, siRNA, nanoparticles, and solid phase surfaces for therapeutic and diagnostic applications. However, as relatively small sized oligonucleotides, aptamers present several challenges for successful clinical translation. Their short plasma half-lives due to nuclease degradation and rapid renal excretion necessitate further structural modification of aptamers for clinical application. Since the US Food and Drug Administration (FDA) approval of the first aptamer drug, Macugen® (pegaptanib), which treats wet-age-related macular degeneration, several aptamer therapeutics for oncology have followed and shown promise in pre-clinical models as well as clinical trials. This review discusses the advantages and challenges of aptamers and introduces therapeutic aptamers under investigation and in clinical trials for cancer treatments.
Collapse
Affiliation(s)
- Yoshihiro Morita
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
| | - Macall Leslie
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
| | - Hiroyasu Kameyama
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
| | - David E Volk
- McGovern Medical School, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston, TX 77030, USA.
| | - Takemi Tanaka
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE 10th, BRC-W, Rm 1415, Oklahoma City, OK 73104, USA.
- Department of Pathology, College of Medicine, University of Oklahoma Health Sciences Center, 940 SL Young Blvd, Oklahoma City, OK 73104, USA.
| |
Collapse
|
20
|
Wolfson E, Solomon S, Schmukler E, Goldshmit Y, Pinkas-Kramarski R. Nucleolin and ErbB2 inhibition reduces tumorigenicity of ErbB2-positive breast cancer. Cell Death Dis 2018; 9:47. [PMID: 29352243 PMCID: PMC5833446 DOI: 10.1038/s41419-017-0067-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/01/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022]
Abstract
ErbB2, a member of the ErbB family of receptor tyrosine kinases, is an essential player in the cell's growth and proliferation signaling pathways. Amplification or overexpression of ErbB2 is observed in ∼30% of breast cancer patients, and often drives cellular transformation and cancer development. Recently, we have shown that ErbB2 interacts with the nuclear-cytoplasmic shuttling protein nucleolin, an interaction which enhances cell transformation in vitro, and increases mortality risk and disease progression rate in human breast cancer patients. Given these results, and since acquired resistance to anti-ErbB2-targeted therapy is a major obstacle in treatment of breast cancer, we have examined the therapeutic potential of targeting the ErbB2-nucleolin complex. The effect of the nucleolin-specific inhibitor GroA (AS1411) on ErbB2-positive breast cancer was tested in vivo, in a mouse xenograft model for breast cancer; as well as in vitro, alone and in combination with the ErbB2 kinase-inhibitor tyrphostin AG-825. Here, we show that in vivo treatment of ErbB2-positive breast tumor xenografts with GroA reduces tumor size and leads to decreased ErbB2-mediated signaling. Moreover, we found that co-treatment of breast cancer cell lines with GroA and the ErbB2 kinase-inhibitor tyrphostin AG-825 enhances the anti-cancer effects exerted by GroA alone in terms of cell viability, mortality, migration, and invasiveness. We, therefore, suggest a novel therapeutic approach, consisting of combined inhibition of ErbB2 and nucleolin, which has the potential to improve breast cancer treatment efficacy.
Collapse
Affiliation(s)
- Eya Wolfson
- Department of Neurobiology, Faculty of Life Science, Tel-Aviv University, Ramat-Aviv, 69978, Israel
| | - Shira Solomon
- Department of Neurobiology, Faculty of Life Science, Tel-Aviv University, Ramat-Aviv, 69978, Israel
| | - Eran Schmukler
- Department of Neurobiology, Faculty of Life Science, Tel-Aviv University, Ramat-Aviv, 69978, Israel
| | - Yona Goldshmit
- Department of Neurobiology, Faculty of Life Science, Tel-Aviv University, Ramat-Aviv, 69978, Israel
| | - Ronit Pinkas-Kramarski
- Department of Neurobiology, Faculty of Life Science, Tel-Aviv University, Ramat-Aviv, 69978, Israel.
| |
Collapse
|
21
|
Abstract
Aptamers are nucleic acids referred to as chemical antibodies as they bind to their specific targets with high affinity and selectivity. They are selected via an iterative process known as ‘selective evolution of ligands by exponential enrichment’ (SELEX). Aptamers have been developed against numerous cancer targets and among them, many tumor cell-membrane protein biomarkers. The identification of aptamers targeting cell-surface proteins has mainly been performed by two different strategies: protein- and cell-based SELEX, when the targets used for selection were proteins and cells, respectively. This review aims to update the literature on aptamers targeting tumor cell surface protein biomarkers, highlighting potentials, pitfalls of protein- and cell-based selection processes and applications of such selected molecules. Aptamers as promising agents for diagnosis and therapeutic approaches in oncology are documented, as well as aptamers in clinical development.
Collapse
|
22
|
Glazyrin YE, Komarova MA, Bakhtina VI, Silacheva MV, Demko IV, Zamay AS, Zamay TN. A comparative protein profiling of lymphocytes from blood of patients with chronic lymphoid leukemia by high-resolution mass-spectrometry in search of new markers for heterogeneity and disease prognosis. JOURNAL OF ANALYTICAL CHEMISTRY 2017. [DOI: 10.1134/s1061934816130062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Malik MT, O'Toole MG, Casson LK, Thomas SD, Bardi GT, Reyes-Reyes EM, Ng CK, Kang KA, Bates PJ. AS1411-conjugated gold nanospheres and their potential for breast cancer therapy. Oncotarget 2016; 6:22270-81. [PMID: 26045302 PMCID: PMC4673162 DOI: 10.18632/oncotarget.4207] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/22/2015] [Indexed: 12/16/2022] Open
Abstract
AS1411 is a quadruplex-forming DNA oligonucleotide that functions as an aptamer to target nucleolin, a protein present on the surface of cancer cells. Clinical trials of AS1411 have indicated it is well tolerated with evidence of therapeutic activity, but improved pharmacology and potency may be required for optimal efficacy. In this report, we describe how conjugating AS1411 to 5 nm gold nanospheres influences its activities in vitro and in vivo. We find that the AS1411-linked gold nanospheres (AS1411-GNS) are stable in aqueous and serum-containing solutions. Compared to unconjugated AS1411 or GNS linked to control oligonucleotides, AS1411-GNS have superior cellular uptake and markedly increased antiproliferative/cytotoxic effects. Similar to AS1411, AS1411-GNS show selectivity for cancer cells compared to non-malignant cells. In a mouse model of breast cancer, systemic administration of AS1411-GNS could completely inhibit tumor growth with no signs of toxicity. These results suggest AS1411-GNS are promising candidates for clinical translation.
Collapse
Affiliation(s)
- Mohammad T Malik
- Departments of Medicine, University of Louisville, Louisville, Kentucky, USA.,Departments of the Molecular Targets Group of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Martin G O'Toole
- Departments of Bioengineering, University of Louisville, Louisville, Kentucky, USA
| | - Lavona K Casson
- Departments of Medicine, University of Louisville, Louisville, Kentucky, USA.,Departments of the Molecular Targets Group of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Shelia D Thomas
- Departments of Medicine, University of Louisville, Louisville, Kentucky, USA.,Departments of the Molecular Targets Group of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Gina T Bardi
- Departments of Medicine, University of Louisville, Louisville, Kentucky, USA.,Departments of the Molecular Targets Group of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Elsa Merit Reyes-Reyes
- Departments of Medicine, University of Louisville, Louisville, Kentucky, USA.,Departments of the Molecular Targets Group of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Chin K Ng
- Departments of Radiology, University of Louisville, Louisville, Kentucky, USA
| | - Kyung A Kang
- Departments of Chemical Engineering, University of Louisville, Louisville, Kentucky, USA
| | - Paula J Bates
- Departments of Medicine, University of Louisville, Louisville, Kentucky, USA.,Departments of the Molecular Targets Group of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
24
|
Aptamers in hematological malignancies and their potential therapeutic implications. Crit Rev Oncol Hematol 2016; 106:108-17. [PMID: 27637356 DOI: 10.1016/j.critrevonc.2016.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/06/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023] Open
Abstract
Aptamers are short DNA/RNA oligonucleotides selected by the process called Systematic Evolution of Ligands by Exponential Enrichment (SELEX). Due to their functional similarity to monoclonal antibodies with some superior characters, such as high specificity and affinity, flexible modification and stability, and lack of toxicity and immunogenicity, they are promising alternative and complementary targeted therapy for hematologic malignancies. The trends in aptamer technology including production, selection, modifications are briefly discussed in this review. The key aspect is to illustrate aptamers against cancer cells in hematologic malignancies especially those that have entered clinical trials. We also discuss some challenges remain in the application of aptamers.
Collapse
|
25
|
Zhu Q, Liu G, Kai M. DNA Aptamers in the Diagnosis and Treatment of Human Diseases. Molecules 2015; 20:20979-97. [PMID: 26610462 PMCID: PMC6332121 DOI: 10.3390/molecules201219739] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
Aptamers have a promising role in the field of life science and have been extensively researched for application as analytical tools, therapeutic agents and as vehicles for targeted drug delivery. Compared with RNA aptamers, DNA aptamers have inherent advantages in stability and facility of generation and synthesis. To better understand the specific potential of DNA aptamers, an overview of the progress in the generation and application of DNA aptamers in human disease diagnosis and therapy are presented in this review. Special attention is given to researches that are relatively close to practical application. DNA aptamers are expected to have great potential in the diagnosis and treatment of human diseases.
Collapse
Affiliation(s)
- Qinchang Zhu
- Faculty of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| | - Ge Liu
- Department of Genomic Epidemiology, Research Center for Environment and Developmental Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan.
| | - Masaaki Kai
- Faculty of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| |
Collapse
|
26
|
Belyaeva TA, Nicol C, Cesur O, Travé G, Blair GE, Stonehouse NJ. An RNA Aptamer Targets the PDZ-Binding Motif of the HPV16 E6 Oncoprotein. Cancers (Basel) 2014; 6:1553-69. [PMID: 25062098 PMCID: PMC4190555 DOI: 10.3390/cancers6031553] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/23/2014] [Accepted: 07/03/2014] [Indexed: 01/01/2023] Open
Abstract
Human papillomavirus 16 (HPV16) is a high-risk DNA tumour virus which is the primary causative agent of cervical cancer. Cell transformation arises from deregulated expression of the E6 and E7 oncogenes. E6 has been shown to bind a number of cellular proteins, including p53 and proteins containing a PDZ domain. This study reports the first RNA aptamers to E6. These have been employed as molecular tools to further investigate E6-p53 and E6-PDZ interactions. This study is focussed on two aptamers (termed F2 and F4) which induced apoptosis in cells derived from an HPV16-transformed cervical carcinoma. The molecules were able to inhibit the interaction between E6 and PDZ1 from Magi1, with F2 being the most effective inhibitor. Neither of the aptamers inhibited E6-p53 interaction or p53 degradation. This study shows the specificity of this approach and highlights the potential benefits of the E6 aptamers as potential therapeutic or diagnostic agents in the future.
Collapse
Affiliation(s)
- Tamara A Belyaeva
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Clare Nicol
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Ozlem Cesur
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Gilles Travé
- UMR 7242 CNRS-Université de Strasbourg, Ecole Supérieure de Biotechnologie, Boulevard Sébastien Brant, Illkirch 67412, France.
| | - George Eric Blair
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Nicola J Stonehouse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
27
|
Xiong X, Lv Y, Chen T, Zhang X, Wang K, Tan W. Nucleic acid aptamers for living cell analysis. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2014; 7:405-426. [PMID: 24896309 DOI: 10.1146/annurev-anchem-071213-015944] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Cells as the building blocks of life determine the basic functions and properties of a living organism. Understanding the structure and components of a cell aids in the elucidation of its biological functions. Moreover, knowledge of the similarities and differences between diseased and healthy cells is essential to understanding pathological mechanisms, identifying diagnostic markers, and designing therapeutic molecules. However, monitoring the structures and activities of a living cell remains a challenging task in bioanalytical and life science research. To meet the requirements of this task, aptamers, as "chemical antibodies," have become increasingly powerful tools for cellular analysis. This article reviews recent advances in the development of nucleic acid aptamers in the areas of cell membrane analysis, cell detection and isolation, real-time monitoring of cell secretion, and intracellular delivery and analysis with living cell models. Limitations of aptamers and possible solutions are also discussed.
Collapse
Affiliation(s)
- Xiangling Xiong
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha 410082, China
| | | | | | | | | | | |
Collapse
|
28
|
Dam DM, Culver KSB, Odom TW. Grafting aptamers onto gold nanostars increases in vitro efficacy in a wide range of cancer cell types. Mol Pharm 2014; 11:580-7. [PMID: 24422969 PMCID: PMC3974612 DOI: 10.1021/mp4005657] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 12/09/2013] [Accepted: 01/14/2014] [Indexed: 02/06/2023]
Abstract
We report the design of a nanoconstruct that can function as a cell-type independent agent by targeting the ubiquitous protein nucleolin. Gold nanostars (AuNS) loaded with high densities of nucleolin-specific DNA aptamer AS1411 (Apt-AuNS) produced anticancer effects in a panel of 12 cancer lines containing four representative subcategories. We found that the nanoconstructs could be internalized by cancer cells and trafficked to perinuclear regions. Apt-AuNS resulted in downregulation of antiapoptotic Bcl-2 mRNA expression by ca. 200% compared to cells without the nanoconstructs. The caspase 3/7 activity (apoptosis) and cell death in cancer cells treated with Apt-AuNS increased by 1.5 times and by ca. 17%, respectively, compared to cells treated with free AS1411 at over 10 times the concentration. Moreover, light-triggered release of aptamer from the AuNS further enhanced the in vitro efficacy of the nanoconstructs in the cancer line panel with a 2-fold increase in caspase activity and a 40% decrease in cell viability compared to treatment with Apt-AuNS only. In contrast, treatments of the nanoconstructs with or without light-triggered release on a panel of normal cell lines had no adverse effects.
Collapse
Affiliation(s)
- Duncan
Hieu M. Dam
- Department
of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Kayla S. B. Culver
- Department
of Materials Science and Engineering, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
| | - Teri W. Odom
- Department
of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Department
of Materials Science and Engineering, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
| |
Collapse
|
29
|
Abstract
Aptamers are single-stranded structured oligonucleotides (DNA or RNA) that can bind to a wide range of targets ("apatopes") with high affinity and specificity. These nucleic acid ligands, generated from pools of random-sequence by an in vitro selection process referred to as systematic evolution of ligands by exponential enrichment (SELEX), have now been identified as excellent tools for chemical biology, therapeutic delivery, diagnosis, research, and monitoring therapy in real-time imaging. Today, aptamers represent an interesting class of modern Pharmaceuticals which with their low immunogenic potential mimic extend many of the properties of monoclonal antibodies in diagnostics, research, and therapeutics. More recently, chimeric aptamer approach employing many different possible types of chimerization strategies has generated more stable and efficient chimeric aptamers with aptamer-aptamer, aptamer-nonaptamer biomacromolecules (siRNAs, proteins) and aptamer-nanoparticle chimeras. These chimeric aptamers when conjugated with various biomacromolecules like locked nucleic acid (LNA) to potentiate their stability, biodistribution, and targeting efficiency, have facilitated the accurate targeting in preclinical trials. We developed LNA-aptamer (anti-nucleolin and EpCAM) complexes which were loaded in iron-saturated bovine lactofeerin (Fe-blf)-coated dopamine modified surface of superparamagnetic iron oxide (Fe3O4) nanoparticles (SPIONs). This complex was used to deliver the specific aptamers in tumor cells in a co-culture model of normal and cancer cells. This review focuses on the chimeric aptamers, currently in development that are likely to find future practical applications in concert with other therapeutic molecules and modalities.
Collapse
Affiliation(s)
- Jagat R Kanwar
- Nanomedicine Laboratory of Immunology and Molecular Biomedical Research (LIMBR), Centre for Biotechnology and Interdisciplinary Biosciences (BioDeakin), Institute for Technology and Research Innovation (ITRI), Geelong Technology Precinct (GTP), Deakin University, Victoria, Australia.
| | | | | |
Collapse
|